GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
  • 1
    Online-Ressource
    Online-Ressource
    Oxford University Press (OUP) ; 2017
    In:  Journal of Leukocyte Biology Vol. 102, No. 4 ( 2017-10-01), p. 1143-1151
    In: Journal of Leukocyte Biology, Oxford University Press (OUP), Vol. 102, No. 4 ( 2017-10-01), p. 1143-1151
    Kurzfassung: Mutations in ELANE, the gene for neutrophil elastase (NE), a protease expressed early in neutrophil development, are the most frequent cause of cyclic (CyN) and severe congenital neutropenia (SCN). We hypothesized that inhibitors of NE, acting either by directly inhibiting enzymatic activity or as chaperones for the mutant protein, might be effective as therapy for CyN and SCN. We investigated β-lactam–based inhibitors of human NE (Merck Research Laboratories, Kenilworth, NJ, USA), focusing on 1 inhibitor called MK0339, a potent, orally absorbed agent that had been tested in clinical trials and shown to have a favorable safety profile. Because fresh, primary bone marrow cells are rarely available in sufficient quantities for research studies, we used 3 cellular models: patient-derived, induced pluripotent stem cells (iPSCs); HL60 cells transiently expressing mutant NE; and HL60 cells with regulated expression of the mutant enzyme. In all 3 models, the cells expressing the mutant enzyme had reduced survival as measured with annexin V and FACS. Coincubation with the inhibitors, particularly MK0339, promoted cell survival and increased formation of mature neutrophils. These studies suggest that cell-permeable inhibitors of neutrophil elastase show promise as novel therapies for ELANE-associated neutropenia.
    Materialart: Online-Ressource
    ISSN: 0741-5400 , 1938-3673
    RVK:
    Sprache: Englisch
    Verlag: Oxford University Press (OUP)
    Publikationsdatum: 2017
    ZDB Id: 2026833-6
    SSG: 12
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Blood, American Society of Hematology, Vol. 139, No. 5 ( 2022-02-03), p. 779-791
    Kurzfassung: Severe congenital neutropenia is an inborn disorder of granulopoiesis. Approximately one third of cases do not have a known genetic cause. Exome sequencing of 104 persons with congenital neutropenia identified heterozygous missense variants of CLPB (caseinolytic peptidase B) in 5 severe congenital neutropenia cases, with 5 more cases identified through additional sequencing efforts or clinical sequencing. CLPB encodes an adenosine triphosphatase that is implicated in protein folding and mitochondrial function. Prior studies showed that biallelic mutations of CLPB are associated with a syndrome of 3-methylglutaconic aciduria, cataracts, neurologic disease, and variable neutropenia. However, 3-methylglutaconic aciduria was not observed and, other than neutropenia, these clinical features were uncommon in our series. Moreover, the CLPB variants are distinct, consisting of heterozygous variants that cluster near the adenosine triphosphate-binding pocket. Both genetic loss of CLPB and expression of CLPB variants result in impaired granulocytic differentiation of human hematopoietic progenitor cells and increased apoptosis. These CLPB variants associate with wild-type CLPB and inhibit its adenosine triphosphatase and disaggregase activity in a dominant-negative fashion. Finally, expression of CLPB variants is associated with impaired mitochondrial function but does not render cells more sensitive to endoplasmic reticulum stress. Together, these data show that heterozygous CLPB variants are a new and relatively common cause of congenital neutropenia and should be considered in the evaluation of patients with congenital neutropenia.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2022
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2005
    In:  Blood Vol. 106, No. 11 ( 2005-11-16), p. 3071-3071
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 3071-3071
    Kurzfassung: Myelokathexis (WHIM syndrome) is a very rare hematopoietic congenital disorder that is characterized by extremely low level of circulating neutrophils in peripheral blood. It is inherited as an autosomal dominant disease and is diagnosed in early childhood. These patients may have hypogammaglobulinemia and suffer from recurrent infections associated with warts. The hallmark of myelokathexis is a hyperplastic bone marrow and hypersegmented neutrophils with nuclear lobs connected with thin filaments. Myelokathexis is due to a characteristic retention of mature neutrophils in bone marrow, which are not being released to peripheral circulation. We and others reported abnormal cell survival characteristics and impaired bcl-x expression in bone marrow myeloid cells of myelokathexis patients that was partially restored by G-CSF treatment. Recently, it has also been reported that heterozygous truncation mutations in the carboxyterminal domain of the CXCR4 gene, a sole receptor for SDF-1 chemokine, were observed in most, but not all of the families with WHIM syndrome. Subsequently, an impaired receptor internalization and increased chemotaxis towards SDF-1 have been observed in cells expressing truncated CXCR4. Nevertheless, the mechanism of mutant CXCR4 induced myelokathexis remains largely unknown. We performed mutational analysis of the CXCR4 gene in 3 unrelated families with myelokathexis and identified a previously reported R334ter truncation mutation in exon 2 in two of the families. In addition, two silent polymorphisms have been identified in exon 2 of the CXCR4 gene in one of these patients. The third family with afflicted mother and son had a new mutation in the CXCR4 carboxyterminal domain, which resulted in deletion of the last 16 amino acids and subsequent frame shift. None of these mutations were observed in healthy volunteers examined. Since the morphological examination by electron microscopy and flow cytometry analysis of bone marrow cells from some of these patients revealed characteristic apoptotic features, we examined the effect of mutant CXCR4 gene expression on survival of human promyelocytic HL-60 cells. Preliminary data demonstrated that human promyelocytic cells transfected with truncated CXCR4 exhibited impaired cell survival characteristics compared with control HL-60 cells transfected with intact CXCR4. The truncated, but not wild type CXCR4 also increased apoptosis in HL-60 cells induced to differentiate along the granulocytic pathway as determined by flow cytometry of annexin V labeled cells. Thus, these data link together the abnormal survival of proliferating and differentiating myeloid cells in WHIM syndrome with mutant CXCR4 expression. Current studies are focused on elucidation of specific signaling pathways mediating mutant CXCR4-triggered myelokathexis.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2005
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3786-3786
    Kurzfassung: Abstract 3786 Mutations in the gene for neutrophil elastase (ELANE) are the cause for the majority of cases of cyclic and congenital neutropenia. More than 50 different mutations of ELANE have been identified in patients with these disorders. It is not known whether specific ELANE mutations predispose patients to the risk of myelodysplasia (MDS) and acute myeloid leukemia (AML). We have investigated the relationships of ELANE mutations to the risk of MDS/AML in a population of 208 patients with cyclic (N=92) and congenital neutropenia (N=116) having ELANE mutations and receiving granulocyte colony-stimulating factor (G-CSF) therapy for up to 22 years. In the congenital group there were 52 different mutations occurring with increasing frequency from exon 1 (2 mutations) to exon 5 (17 different mutations). In the cyclic group there was a narrower spectrum of mutations; these mutations clustered in exon 4, intron 4 and exon 5. There were 13 different mutations in this group. Mutations P110L, S97L, R191Q, and splice site mutations in intron 4: +5 SD G 〉 A, and +1 SD G 〉 T were observed both in patients with the cyclic and congenital neutropenia. Sixteen congenital neutropenic patients developed MDS/AML. There were 15 different mutations associated with these transformations. There are also 15 congenital patients who have not evolved to develop leukemia who have the same mutations as in the MDS/AML group. In three families with autosomal dominant congenital neutropenia attributed to ELANE mutations, one affected member has developed AML the other has not. The pairs are: father no, daughter yes; father yes, daughter no; and one identical twin yes, other twin no. One patient with a clinical diagnosis of cyclic neutropenia who had received long term immunosuppressive therapy also developed MDS and had a stem cell transplant. There were no MDS/AML transformations in the other 91 cyclic patients. For the overall population of cyclic and congenital patients, missense mutations in ELANE were most commonly found. In the 16 congenital neutropenia patients evolving to MDS/AML by category the mutations were: missense (8), deletion (2) termination (4) intronic (2), a pattern similar to those of the congenital patients who have not evolved to develop MDS/AML: missense (79) deletion (7) termination (8) intronic (6). The 16 patients with congenital neutropenia who developed MDS/AML were treated with G-CSF for a median of 8.6 years (range 3.3 – 15.2). The mean of the median G-CSF dose for patients with MDS/AML transformation was 14.6 mcg/kg/day (range 2–43 mcg/kg/day) and the mean of the median dose of G-CSF for the population not developing MDS/AML was 13.7 mcg/kg/day (range 0–183 mcg/kg/day) (p=0.89). However, a higher proportion of the AML group (69%) were receiving G-CSF at 〉 8mcg/kg than the non MDS/AML group (40%). The median duration of G-CSF treatment was significantly less (p=0.009) in the MDS/AML group than the group not developing MDS/AML. The mean cumulative G-CSF exposure was not significantly different (p=0.54) for the two groups. These data indicate that there are no specific ELANE mutations or specific types of ELANE mutations predisposing patients with congenital neutropenia to development of MDS or AML. The data confirm previous reports that patients requiring higher daily doses of G-CSF may be at somewhat greater risk of MDS/AML, but there is no distinct dose predicting risk and no association with total G-CSF exposure and the risk of MDS/AML. The specific cause for the high risk of MDS and AML in congenital neutropenia remains unknown. Disclosures: Dale: Amgen: Consultancy, Research Funding.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2010
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 386-386
    Kurzfassung: Abstract 386 Heterozygous mutations in the neutrophil elastase gene ELANE have been identified as the primary cause of severe congenital neutropenia (SCN) associated with recurring severe infections and evolution to acute myeloid leukemia (AML). As of today, more than 50 substitution, truncation, insertion and deletion mutations have been identified. Animal studies based on knock-in or knockout of ELANE in mice failed to produce severe neutropenia phenotype. We and others previously reported that expression of various mutants but not wild type neutrophil elastase (NE) in human but not murine cells triggers accelerated apoptosis. We also reported that expression of mutant NE (del.145-152), identified in SCN patients one of whom evolved to develop MDS/AML, in human promyelocytic tet-off HL60 cells causes both accelerated apoptosis and characteristic block of myeloid differentiation similar to that seen in bone marrow of SCN patients. Examination of the tertiary structure of NE revealed that most of the mutations leave the active site of the mutant protease intact. We identified a small molecule inhibitor of neutrophil elastase, a derivative of L-malic acid (Merck, USA), that blocked the proteolytic activity of NE by approximately 80% and was capable of restoring impaired myeloid differentiation and normalizing production of myeloid cells expressing del145-152 NE mutant. It is important to note that block of proteolytic activity of NE with the NE-SMI had no adverse effect on control human myeloid progenitor cells expressing wild type NE, thus confirming the gain-of-function effect of NE mutants. More than 20% of SCN patients with NE mutations evolve to develop AML. Molecular modeling and analysis of the tertiary structures of NE available through the Protein Database revealed that 16 different mutations identified in AML patients affect predominantly the N95 or N144 glycosylation sites or the binding pocket of the protease suggesting that altered substrate specificity of the mutant enzyme is the cause of accelerated apoptosis and block of myeloid differentiation in SCN/AML. We sought to obtain bone marrow samples from 2 unrelated SCN/AML patients both on G-CSF treatment harboring either C122Y or insPQ94. Bone marrow purified CD34+ and/or CD34-/CD33+ myeloid progenitors from the patients showed basal level of apoptosis in a range of 20–25%, which gradually increased reaching 40–50% apoptosis by 3 days of culture. Importantly, treatment of primary bone marrow-derived cells with NE-SMI substantially reduced accelerated apoptosis to near initial rate with approximately up to 2-fold reduction of apoptosis by 3 days of culture as determined by flow cytometry. Thus, our findings demonstrate that 1) small molecule inhibitor of neutrophil elastase is effective in blocking accelerated apoptosis triggered by three different NE mutations identified in SCN patients evolved to develop MDS/AML and 2) the small molecule inhibitor of NE is a promising therapeutic agent that should be considered for testing in clinical trials in SCN/AML patients. Disclosures: Dale: Amgen: Consultancy, Research Funding; Merck: Patents & Royalties, Research Support.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2010
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2011
    In:  Blood Vol. 118, No. 21 ( 2011-11-18), p. 1105-1105
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 1105-1105
    Kurzfassung: Abstract 1105 Barth syndrome (BTHS) is a rare, X-linked recessive disease characterized by progressive cardiomyopathy and neutropenia with life-threatening bacterial infections. BTHS is attributable to loss-of-function mutations in the tafazzin gene, TAZ, a phospholipid acyltransferase localized to the mitochondrial membrane and involved in remodeling cardiolipin. BTHS children exhibit reduced blood levels of total cardiolipin; however, the molecular and cellular mechanisms of BTHS remain largely unknown. We developed a model of BTHS by transfecting human HL60 myeloid progenitor cells with TAZ-specific shRNAs. Results demonstrated a significant down-regulation in the TAZ gene expression, mimicking naturally-occurring truncation mutations. FACS analyses of cells with TAZ-specific but not scrambled shRNAs demonstrated nearly two-fold increase in proportion of annexin-V positive cells and significantly increased dissipation of mitochondrial membrane potential (MMP) as determined by DIOC6-staining. Similar pro-apoptotic effects were observed in U937 myeloid progenitors, but not in Jurkat or Ramos human lymphoid cell lines. Further studies revealed aberrant release of cytochrome c from mitochondria and significantly elevated levels of activated caspase-3 in response to TAZ knock-down. Treatment with caspase-specific inhibitor zVAD-fmk reduced apoptosis to near-normal levels. FACS analysis utilizing cryopreserved bone marrow cells from a BTHS patient showed evidence of accelerated apoptosis compared to corresponding cells from a healthy volunteer. Importantly, human myeloid progenitor cells and cardiomyoblasts/cardiomyocytes depend heavily on mitochondria, which may explain why the loss of mitochondrial protein tafazzin has more severe effects on these specific tissues in BTHS resulting in neutropenia and cardiomyopathy. These data suggest that neutropenia in BTHS patients is attributable to increased dissipation of MMP, aberrant release of cytochrome c, activation of caspase-3 and accelerated apoptosis, and that this defect can be partially restored in vitro by treatment with caspase-specific inhibitors. Further studies should evaluate the TAZ–regulated mitochondrial apoptosis pathway using primary cells from BTHS patients. Disclosures: Aprikyan: Barth Syndrome Foundation: Research Funding. Dale:Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2011
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 237-237
    Kurzfassung: Abstract 237 Myelokathexis (MK), also known as WHIM syndrome, is a rare congenital autosomal dominant stem cell disorder characterized by aberrant retention of hematopoietic cells in the bone marrow and severe neutropenia and lymphopenia. MK patients may evolve to develop Acute Myeloid Leukemia (AML) or fatal B-cell lymphoma. We and others reported that severe leukopenia in MK is due to accelerated apoptosis of bone marrow CD34+ stem cells and CD33+ myeloid progenitors. Impaired cell survival was associated with reduced Bcl-X expression, which, at least partially is restored by treatment of patients with G-CSF. Most of MK patients harbor heterozygous mutations in the CXCR4 gene product, which interacts with SDF-1 ligand and plays a key role in controlling homing and mobilization of hematopoietic stem cells. Mutant CXCR4 exhibits reduced internalization, enhanced calcium flux and results in elevated chemotaxis of human CD34+ stem cells toward SDF-1. However, the mechanisms of mutant CXCR4-induced severe neutropenia in MK and its malignant evolution remain unknown. We examined 11 patients from 6 unrelated families affected with MK and identified heterozygous CXCR4 mutations in these patients. Expression of different CXCR4 mutants in human HL-60 myeloid progenitor cells cultured in the presence of FBS triggers accelerated apoptosis comparable to that observed in MK. Particularly, the rate of apoptotic annexin V-positive cells was ∼2-fold higher in mutant CXCR4-expressing cells compared with control cells transfected with wild type CXCR4 (24±2% for wt CXCR4 vs 50±6% and 51±5% for different CXCR4 mutants). Similar increase in apoptosis was observed regardless whether the cells were cultured in the presence or absence of serum, SDF-1 or G-CSF, thus suggesting that increased apoptosis of bone marrow cells in MK is due to intrinsic CXCR4-activated pro-apoptotic signal transduction pathway. Accelerated apoptosis appeared to stem from enhanced dissipation of mitochondrial membrane potential as determined by FACS analysis of DIOC6-labeled cells expressing mutant CXCR4 compared with control CXCR4 (p 〈 0.02). Similar to enhanced chemotaxis of patients' mononuclear cells, expression of CXCR4 mutants, but not the normal CXCR4 triggered a robust increase in directional motility of the cells to SDF-1 (p 〈 0.01). These data indicate that this cellular model with mutant CXCR4 expression appears to closely recapitulate the MK phenotype with accelerated apoptosis and impaired mobilization of bone marrow stem cells and myeloid progenitors. Accelerated apoptosis, but not enhanced chemotaxis triggered by mutant CXCR4 was normalized by treatment with caspase-specific inhibitor zVAD-fmk. Interestingly, the mutant CXCR4-induced increase in chemotaxis was restored with treatment with protein kinase C (PKC) inhibitor. The observed normalization of mutant CXCR4-triggerred increase in chemotaxis in response to PKC inhibitor was similar to that observed in response to a treatment with CXCR4 antagonist AMD3100. Interestingly, combined treatment with both PKC inhibitor and AMD3100 had no synergistic effect, suggesting that these compounds may utilize the same signaling pathway. Noteworthy, neither the treatment with AMD3100 nor with PKC inhibitor affected apoptosis, suggesting that observed accelerated apoptosis and increased chemotaxis in MK are two independent pathways triggered by CXCR4 mutants. Importantly, treatment of patients' blood mononuclear cells with PKC inhibitor restored the abnormal chemotactic properties to near normal levels. Thus, our data suggest that PKC inhibitor and AMD3100 may be effective for treatment of MK patients and for controlling the aberrant retention of bone marrow stem and myeloid cells in this stem cell disorder. Disclosures: Dale: Amgen: Consultancy, Research Funding, Speaker; Genzyme: Research support pending.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2009
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2004
    In:  Blood Vol. 104, No. 11 ( 2004-11-16), p. 1453-1453
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 1453-1453
    Kurzfassung: Severe congenital neutropenia (SCN) or Kostmann’s syndrome defines an inheritable hematopoietic disorder of an impaired neutrophil production in the bone marrow due to a “maturation arrest” at promyelocytic stage of differentiation in the marrow. SCN patients have recurring severe infections and may evolve to develop leukemia. We reported accelerated apoptosis and cell cycle arrest of bone marrow-derived myeloid progenitor cells in SCN patients with acquired, autosomal dominant, as well as autosomal recessive inheritance. We also reported that approximately 80% of these patients have heterozygous mutations in the neutrophil elastase (NE) gene encoding a serine protease normally targeted to azurophil granules. Neither knock-in of mutant elastase nor a knock-out of normal neutrophil elastase caused severe neutropenia in mice. Therefore, we established a cellular model of SCN with tetracycline-regulated expression of mutant NE in human promyelocytic tet-off HL-60 cells. Induced expression of mutant elastase in these cells led to a reduced mitochondrial membrane potential and subsequent caspase-independent apoptosis and cell cycle arrest as determined by flow cytometry. Block of differentiation of DMSO-treated cells was also observed in tet-off HL-60 cells with induced expression of mutant NE, similar to that in SCN patients. This cellular model of SCN very closely recapitulates the human severe neutropenia phenotype. To elucidate the molecular mechanisms of mutant NE-mediated neutropenia, we employed various proteomics methods, including ICAT analysis, two-dimensional gel electrophoresis, immunoprecipitation, LC-MS/MS and identified a number of proteins with altered level of expression including apoptosis and cell cycle regulatory gene products, transcription factor and cytoskeleton proteins. Changes in the protein expression profiles revealed the abnormal molecular events underlying the impaired cell survival and cell cycle arrest and suggested additional pathways implicated in pathogenesis of SCN. Immunostaining of control cells with phalloidin revealed a weak F-actin polymerization in cell periphery, which helps to maintain cell shape flexibility, and intracellular long F-actin filaments supporting the normal cytoskeleton. In contrast, cells expressing mutant NE, exhibited an increased polymerization of F-actin in the periphery, with apparent increase in lamellipodia that may contribute to an increased phosphatydilserine exposure in apoptotic cells expressing mutant elastase. The hyperpolymerization of F-actin, which appears to stem from an elevated level of RhoA, a member of the RAS-superfamily of GTPases, makes the cells more rigid and less flexible thus contributing to impaired cell cycle progression in cells expressing mutant NE. Impaired cell survival in these cells is associated with a significant reduction in the level of phosphorylated PKB/Akt, which in turn appears to be a consequence of a decreased level of PI3kinase in response to expression of mutant NE. Interestingly, G-CSF treatment of mutant NE expressing cells resulted in restoration of the level of phospho-Akt to near normal level comparable to that in control cells with normal NE expression. These data may explain the anti-apoptotic effect of G-CSF in SCN. Thus, these data demonstrate that the cellular model of SCN based on tet-off HL-60/mutNE cells with inducible expression of mutant elastase is useful to unravel the cellular and molecular mechanisms of mutant NE-mediated severe neutropenia.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2004
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2008
    In:  Blood Vol. 112, No. 11 ( 2008-11-16), p. 3558-3558
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3558-3558
    Kurzfassung: Barth syndrome (BTHS) is an X-linked recessive disorder characterized by neutropenia, cardiac and skeletal myopathies, and growth retardation. The mortality is high due to progressive cardiomyopathy and/or overwhelming bacterial infections. The incidence of BTHS is estimated to be as high as 1 in 100,000, but it is still a poorly recognized disease. The majority of Barth patients have mutations in the tafazzin (G4.5 or TAZ) gene, most of which appear to truncate the tafazzin protein likely resulting in the loss of its function. Tafazzin is a phospholipid acyltransferase involved in remodeling cardiolipin, the main lipid of the inner mitochondrial membrane. As a result, BTHS patients exhibit reduced levels of total CL and accumulation of monolysocardiolipin. A drosophila model of the Barth syndrome was recently reported, but cellular or mouse models of this disorder are not yet available. The link between these metabolic defects triggered by TAZ mutations and neutropenia remains largely unknown. We hypothesized that TAZ mutations lead to the loss of function of tafazzin protein causing impaired cell survival of neutrophil precursors, reduced production of neutrophils and neutropenia. To test this hypothesis, we knocked down the expression of the tafazzin gene in human myeloid progenitor HL60 cells using TAZ-specific shRNA and examined its effect on cell survival. Four shRNAs specific to exons 4 through 7 of the TAZ gene were used for transfection of human myeloid progenitor HL60 cells that were later examined by flow cytometry and Western blot analyses. At least 2 of the shRNA constructs resulted in substantial down-regulation in the expression level of the tafazzin gene in transfected human myeloid progenitor cells as determined by Western blot and confirmed by RT-PCR using gene-specific and GAPDH-specific primers. Flow cytometry analysis of DIOC6-labeled cells revealed that knock-down of the TAZ gene expression was associated with a significantly elevated dissipation of mitochondrial membrane potential compared with control cells with scrambled shRNA ((p 〈 0.0009, n=3). Apoptosis studies using flow cytometry analyses revealed a significant increase in proportion of apoptotic annexin-positive cells compared with control cells transfected with scrambled shRNA (p 〈 0.0002, n=6). The observed increase in apoptosis in response to TAZ knock-down was caspase3-dependent as evidenced by Western blot analysis. Treatment of the cells with caspase-specific inhibitor zVAD-fmk significantly improved cell survival characteristics to near normal level as determined by flow cytometry (p 〈 0.02, n=4), suggesting that caspase-specific inhibitors may represent potentially therapeutic agents for patients with Barth syndrome. Thus, these data demonstrate that the loss of function of the TAZ gene is cytotoxic to hematopoietic cells and suggest that severe neutropenia is due to accelerated apoptosis of myeloid progenitor cells.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2008
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2009
    In:  Blood Vol. 114, No. 22 ( 2009-11-20), p. 549-549
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 549-549
    Kurzfassung: Abstract 549 Barth syndrome (BTHS) is a severe X-linked stem cell disorder characterized by neutropenia, cardio- and skeletal myopathies, and growth retardation. Barth patients have a high rate of mortality due to progressive cardiomyopathy and/or overwhelming bacterial infections. Majority of Barth patients have mutations in the tafazzin (G4.5 or TAZ) gene that appear to truncate the tafazzin protein resulting in the loss of TAZ function. Based on protein homology, tafazzin is a phospholipid acyltransferase involved in remodeling cardiolipin (CL), the main lipid of the inner mitochondrial membrane. Therefore, BTHS patients exhibit reduced levels of total CL and accumulation of monolysocardiolipin. However, the function of TAZ protein and how these metabolic defects are triggered by TAZ mutations in BTHS remain largely unknown. The cellular or mouse models of this disorder are not availabel yet, and thus, the link between TAZ mutations and severe neutropenia in Barth syndrome remains elusive. Earlier study reported increased annexin V staining but absence of apoptosis in peripheral blood neutrophils. However, the patients' bone marrow stem and myeloid progenitor cells have not been examined. We hypothesized that TAZ mutations trigger accelerated apoptosis of bone marrow stem/progenitor cells, which in turn leads to reduced production of neutrophils in the bone marrow and severe neutropenia in Barth patients. To test this hypothesis, we used TAZ-specific shRNA to knock down the expression of the tafazzin gene in human myeloid progenitor HL60 cells and examined its effect on cell survival. Transfection of human myeloid progenitor cells with two different TAZ-specific but not control scrambled shRNA results in substantial down-regulation in the tafazzin expression level as determined by Western blot and confirmed by RT-PCR using TAZ and GAPDH-specific primers. Human myeloid progenitor cells with knocked down TAZ expression exhibit significantly elevated dissipation of mitochondrial membrane potential compared with control cells with scrambled shRNA as evidenced by flow cytometry analysis of DIOC6-labeled cells ((p 〈 0.0009, n=3). A remarkably significant increase in proportion of apoptotic annexin-positive cells was also observed in response to knock-down of TAZ expression in myeloid progenitor cells compared with control cells with scrambled shRNA (p 〈 0.0002, n=6). The observed increase in apoptosis in response to TAZ knock-down was caspase-3 dependent as evidenced by Western blot analysis. Treatment of the cells with caspase-specific inhibitor zVAD-fmk significantly improved cell survival characteristics to near normal level as determined by flow cytometry (p 〈 0.02, n=4). Interestingly, knock-down of TAZ expression in human lymphoid cells failed to affect their cell survival or mitochondrial membrane potential, indicating that the loss of TAZ function exhibits lineage-specific effect. Analysis of bone marrow-derived CD34+ stem cells from a Barth patient positive for TAZ mutation cultured 24h in the presence of 10% autologous serum revealed nearly 3-fold increase in proportion of apoptotic annexin V positive CD34+ cells compared with the same cell subpopulation from a healthy volunteer (36% in Barth vs 13% in ctrl). More differentiated CD15+ neutrophil precursors also exhibit substantially increased rate of apoptosis compared with control (Barth 27% vs ctrl 17%). CD33+ myeloid-committed progenitor cells from Barth patient do not show increased annexin V binding compared with corresponding cell subpopulation from a healthy volunteer, but demonstrate approximately 2-fold increase in dissipation of mitochondrial membrane potential compared with control donor cells. Thus, these data demonstrate that tafazzin functions as an anti-apoptotic protein, the loss of function of the TAZ gene is cytotoxic to hematopoietic cells, and that severe neutropenia in patients with Barth syndrome is due to accelerated apoptosis of bone marrow myeloid progenitor cells. Our data also reveal that caspase-specific inhibitors may represent potentially therapeutic agents capable of restoring normal production of myeloid cells in Barth Syndrome. Disclosures: No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2009
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...